Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Mech Ageing Dev ; 200: 111583, 2021 12.
Article in English | MEDLINE | ID: mdl-34637937

ABSTRACT

Despite continued increases in human life expectancy, the factors determining the rate of human biological aging remain unknown. Without understanding the molecular mechanisms underlying aging, efforts to prevent aging are unlikely to succeed. The tumor suppression theory of aging introduced here proposes somatic mutation as the proximal cause of aging, but postulates that oncogenic transformation and clonal expansion, not functional impairment, are the relevant consequences of somatic mutation. Obesity and caloric restriction accelerate and decelerate aging due to their effect on cell proliferation, during which most mutations arise. Most phenotypes of aging are merely tumor-suppressive mechanisms that evolved to limit malignant growth, the dominant age-related cause of death in early and middle life. Cancer limits life span for most long-lived mammals, a phenomenon known as Peto's paradox. Its conservation across species demonstrates that mutation is a fundamental but hard limit on mammalian longevity. Cell senescence and apoptosis and differentiation induced by oncogenes, telomere shortening or DNA damage evolved as a second line of defense to limit the tumorigenic potential of clonally expanding cells, but accumulating senescent cells, senescence-associated secretory phenotypes and stem cell exhaustion eventually cause tissue dysfunction and the majority, if not most, phenotypes of aging.


Subject(s)
Aging/physiology , Carcinogenesis , Cell Self Renewal/physiology , Clonal Evolution/physiology , Longevity/physiology , Caloric Restriction , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Transformation, Neoplastic , Humans , Mutation Accumulation
2.
Adv Cancer Res ; 151: 39-67, 2021.
Article in English | MEDLINE | ID: mdl-34148619

ABSTRACT

Multi-modal treatment of non-metastatic locally advanced rectal adenocarcinoma (LARC) includes chemotherapy, radiation, and life-altering surgery. Although highly effective for local cancer control, metastatic failure remains significant and drives rectal cancer-related mortality. A consistent observation of this tri-modality treatment paradigm is that histologic response of the primary tumor to neoadjuvant treatment(s), which varies across patients, predicts overall oncologic outcome. In this chapter, we will examine this treatment response heterogeneity in the context of evolutionary dynamics. We hypothesize that improved understanding of eco-evolutionary pressures rendering small cancer cell populations vulnerable to extinction may influence treatment strategies and improve patient outcomes. Applying effective treatment(s) to cancer populations causes a "race to extinction." We explore principles of eco-evolutionary extinction in the context of these small cancer cell populations, evaluating how treatment(s) aim to eradicate the cancer populations to ultimately result in cure. In this chapter, we provide an evolutionary rationale for limiting continuous treatment(s) with the same agent or combination of agents to avoid selection of resistant cancer subpopulation phenotypes, allowing "evolutionary rescue." We draw upon evidence from nature demonstrating species extinction rarely occurring as a single event phenomenon, but rather a series of events in the slide to extinction. We posit that eradicating small cancer populations, similar to small populations in natural extinctions, will usually require a sequence of different external perturbations that produce negative, synergistic dynamics termed the "extinction vortex." By exploiting these unique extinction vulnerabilities of small cancer populations, the optimal therapeutic sequences may be informed by evolution-informed strategies for patients with LARC.


Subject(s)
Adenocarcinoma/pathology , Clonal Evolution/physiology , Neoadjuvant Therapy/adverse effects , Rectal Neoplasms/pathology , Adaptation, Physiological/drug effects , Adaptation, Physiological/radiation effects , Adenocarcinoma/therapy , Animals , Chemotherapy, Adjuvant/adverse effects , Clonal Evolution/drug effects , Clonal Evolution/radiation effects , Disease Progression , Humans , Radiotherapy/adverse effects , Rectal Neoplasms/therapy
4.
Cancer Res ; 81(4): 1123-1134, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33293425

ABSTRACT

Cancer screening and early detection efforts have been partially successful in reducing incidence and mortality, but many improvements are needed. Although current medical practice is informed by epidemiologic studies and experts, the decisions for guidelines are ultimately ad hoc. We propose here that quantitative optimization of protocols can potentially increase screening success and reduce overdiagnosis. Mathematical modeling of the stochastic process of cancer evolution can be used to derive and optimize the timing of clinical screens so that the probability is maximal that a patient is screened within a certain "window of opportunity" for intervention when early cancer development may be observable. Alternative to a strictly empirical approach or microsimulations of a multitude of possible scenarios, biologically based mechanistic modeling can be used for predicting when best to screen and begin adaptive surveillance. We introduce a methodology for optimizing screening, assessing potential risks, and quantifying associated costs to healthcare using multiscale models. As a case study in Barrett's esophagus, these methods were applied for a model of esophageal adenocarcinoma that was previously calibrated to U.S. cancer registry data. Optimal screening ages for patients with symptomatic gastroesophageal reflux disease were older (58 for men and 64 for women) than what is currently recommended (age > 50 years). These ages are in a cost-effective range to start screening and were independently validated by data used in current guidelines. Collectively, our framework captures critical aspects of cancer evolution within patients with Barrett's esophagus for a more personalized screening design. SIGNIFICANCE: This study demonstrates how mathematical modeling of cancer evolution can be used to optimize screening regimes, with the added potential to improve surveillance regimes. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/4/1123/F1.large.jpg.


Subject(s)
Early Detection of Cancer/methods , Models, Theoretical , Population Surveillance/methods , Adenocarcinoma/diagnosis , Adenocarcinoma/epidemiology , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Barrett Esophagus/diagnosis , Barrett Esophagus/epidemiology , Barrett Esophagus/pathology , Calibration , Clonal Evolution/physiology , Cost-Benefit Analysis , Datasets as Topic , Early Detection of Cancer/economics , Early Detection of Cancer/standards , Esophageal Neoplasms/diagnosis , Esophageal Neoplasms/epidemiology , Esophageal Neoplasms/pathology , Female , Gastroesophageal Reflux/diagnosis , Gastroesophageal Reflux/epidemiology , Gastroesophageal Reflux/pathology , Humans , Incidence , Male , Mass Screening/economics , Mass Screening/methods , Mass Screening/standards , Middle Aged , United States/epidemiology
5.
Nat Rev Mol Cell Biol ; 21(12): 715-728, 2020 12.
Article in English | MEDLINE | ID: mdl-32968234

ABSTRACT

In the 20 years since human embryonic stem cells, and subsequently induced pluripotent stem cells, were first described, it has become apparent that during long-term culture these cells (collectively referred to as 'pluripotent stem cells' (PSCs)) can acquire genetic changes, which commonly include gains or losses of particular chromosomal regions, or mutations in certain cancer-associated genes, especially TP53. Such changes raise concerns for the safety of PSC-derived cellular therapies for regenerative medicine. Although acquired genetic changes may not be present in a cell line at the start of a research programme, the low sensitivity of current detection methods means that mutations may be difficult to detect if they arise but are present in only a small proportion of the cells. In this Review, we discuss the types of mutations acquired by human PSCs and the mechanisms that lead to their accumulation. Recent work suggests that the underlying mutation rate in PSCs is low, although they also seem to be particularly susceptible to genomic damage. This apparent contradiction can be reconciled by the observations that, in contrast to somatic cells, PSCs are programmed to die in response to genomic damage, which may reflect the requirements of early embryogenesis. Thus, the common genetic variants that are observed are probably rare events that give the cells with a selective growth advantage.


Subject(s)
Clonal Evolution/genetics , Mutation Accumulation , Pluripotent Stem Cells/metabolism , Cell Culture Techniques/methods , Cell Culture Techniques/standards , Cell Differentiation/genetics , Cell- and Tissue-Based Therapy/methods , Cell- and Tissue-Based Therapy/trends , Cells, Cultured , Clonal Evolution/physiology , Human Embryonic Stem Cells/physiology , Humans , Mutation/physiology , Pluripotent Stem Cells/physiology
6.
Br J Cancer ; 123(10): 1562-1569, 2020 11.
Article in English | MEDLINE | ID: mdl-32848201

ABSTRACT

BACKGROUND: Tumour hypoxia is associated with metastatic disease, and while there have been many mechanisms proposed for why tumour hypoxia is associated with metastatic disease, it remains unclear whether one precise mechanism is the key reason or several in concert. Somatic evolution drives cancer progression and treatment resistance, fuelled not only by genetic and epigenetic mutation but also by selection from interactions between tumour cells, normal cells and physical micro-environment. Ecological habitats influence evolutionary dynamics, but the impact on tempo of evolution is less clear. METHODS: We explored this complex dialogue with a combined clinical-theoretical approach by simulating a proliferative hierarchy under heterogeneous oxygen availability with an agent-based model. Predictions were compared against histology samples taken from glioblastoma patients, stained to elucidate areas of necrosis and TP53 expression heterogeneity. RESULTS: Results indicate that cell division in hypoxic environments is effectively upregulated, with low-oxygen niches providing avenues for tumour cells to spread. Analysis of human data indicates that cell division is not decreased under hypoxia, consistent with our results. CONCLUSIONS: Our results suggest that hypoxia could be a crucible that effectively warps evolutionary velocity, making key mutations more likely. Thus, key tumour ecological niches such as hypoxic regions may alter the evolutionary tempo, driving mutations fuelling tumour heterogeneity.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Clonal Evolution/physiology , Glioblastoma/genetics , Glioblastoma/pathology , Tumor Hypoxia/physiology , Algorithms , Brain Neoplasms/metabolism , Cell Hypoxia/physiology , Cell Line, Tumor , Cell Proliferation/genetics , Computational Biology/methods , Disease Progression , Glioblastoma/metabolism , High-Throughput Nucleotide Sequencing/methods , High-Throughput Nucleotide Sequencing/statistics & numerical data , Humans , Image Processing, Computer-Assisted/methods , Image Processing, Computer-Assisted/statistics & numerical data , Models, Theoretical , Neoplasm Metastasis , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Oxygen/metabolism , Time Factors
7.
Annu Rev Pathol ; 15: 419-438, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31689371

ABSTRACT

Traditional risk factors are incompletely predictive of cardiovascular disease development, a leading cause of death in the elderly. Recent epidemiological studies have shown that human aging is associated with an increased frequency of somatic mutations in the hematopoietic system, which provide a competitive advantage to a mutant cell, thus allowing for its clonal expansion, a phenomenon known as clonal hematopoiesis. Unexpectedly, these mutations have been associated with a higher incidence of cardiovascular disease, suggesting a previously unrecognized connection between somatic mutations in hematopoietic cells and cardiovascular disease. Here, we provide an up-to-date review of clonal hematopoiesis and its association with aging and cardiovascular disease. We also give a detailed report of the experimental studies that have been instrumental in understanding the relationship between clonal hematopoiesis and cardiovascular disease and have shed light on the mechanisms by which hematopoietic somatic mutations contribute to disease pathology.


Subject(s)
Aging/physiology , Cardiovascular Diseases/etiology , Clonal Evolution/physiology , Hematopoiesis/physiology , Aged , Aged, 80 and over , Aging/genetics , Cardiovascular Diseases/epidemiology , Cells, Cultured , Hematopoiesis/genetics , Humans , Incidence , Mutation/physiology , Risk Factors
8.
Sci Rep ; 9(1): 17380, 2019 11 22.
Article in English | MEDLINE | ID: mdl-31758030

ABSTRACT

Tumor clonal heterogeneity drives treatment resistance. But robust models are lacking that permit eavesdropping on the basic interaction network of tumor clones. We developed an in vitro, functional model of clonal cooperation using U87MG glioblastoma cells, which isolates fundamental clonal interactions. In this model pre-labeled clones are co-cultured to track changes in their individual motility, growth, and drug resistance behavior while mixed. This highly reproducible system allowed us to address a new class of fundamental questions about clonal interactions. We demonstrate that (i) a single clone can switch off the motility of the entire multiclonal U87MG cell line in 3D culture, (ii) maintenance of clonal heterogeneity is an intrinsic and influential cancer cell property, where clones coordinate growth rates to protect slow growing clones, and (iii) two drug sensitive clones can develop resistance de novo when cooperating. Furthermore, clonal communication for these specific types of interaction did not require diffusible factors, but appears to depend on cell-cell contact. This model constitutes a straightforward but highly reliable tool for isolating the complex clonal interactions that make up the fundamental "hive mind" of the tumor. It uniquely exposes clonal interactions for future pharmacological and biochemical studies.


Subject(s)
Brain Neoplasms/pathology , Cell Movement , Cell Proliferation , Clone Cells/pathology , Drug Resistance, Neoplasm , Glioblastoma/pathology , Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Communication/drug effects , Cell Communication/physiology , Cell Line, Tumor , Clonal Evolution/physiology , Clone Cells/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Profiling , Genetic Heterogeneity , Genotype , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Models, Biological , Signal Transduction/genetics
9.
JAMA Cardiol ; 4(4): 380-387, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30865214

ABSTRACT

Importance: Clonal hematopoiesis (CH) has been recently described as a novel driver for cancer and cardiovascular disease (CVD). Clonal hematopoiesis is a common, age-associated disorder marked by expansion of hematopoietic clones carrying recurrent somatic mutations. Current literature suggests that patients with CH have a higher risk of subsequent hematological malignant conditions and mortality attributable to excess CVD. This review discusses the association of cancer with CVD with CH as a potential unifying factor. Observations: The prevalence of CH varies based on the sequencing depth, diagnostic criteria, and patient age and ranges from less than 1% in those younger than 40 years to more than 15% to 20% in those 90 years and older. Clonal hematopoiesis is associated with a 0.5% to 1.0% absolute annual risk of hematological malignant condition and a 2-fold to 4-fold higher risk of coronary artery disease, stroke, and CVD deaths, independent of traditional cardiovascular risk factors. In fact, CH appears to have a relative risk similar to that of traditional cardiovascular risk factors for CVD. Experimental studies suggest that the link between CVD and CH is causal, with inflammation as 1 potential mechanism. There may be also a link between CH and CVD in survivors of cancer; however, data to support this association are currently limited. Conclusions and Relevance: Clonal hematopoiesis represents a premalignant state, with carriers having an increased risk of hematological malignant conditions. Although most carriers will not develop a malignant condition, CH confers an increased risk of CVD, possibly via inflammation. Clonal hematopoiesis may also contribute to CVD in survivors of cancer, although this hypothesis requires validation. Clinically, as advanced sequencing techniques become available, CH may pave the way for precision medicine in the field of cardio-oncology.


Subject(s)
Cardiovascular Diseases/genetics , Clonal Evolution/physiology , Hematopoiesis/genetics , Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/mortality , Coronary Artery Disease/genetics , Coronary Artery Disease/mortality , Humans , Inflammation/genetics , Middle Aged , Mutation/genetics , Neoplasms/epidemiology , Neoplasms/mortality , Prevalence , Risk Factors , Stroke/epidemiology , Stroke/genetics , Stroke/mortality
10.
Nat Commun ; 9(1): 3535, 2018 08 30.
Article in English | MEDLINE | ID: mdl-30166543

ABSTRACT

Pre-leukemic stem cells (pre-LSCs) give rise to leukemic stem cells through acquisition of additional gene mutations and are an important source of relapse following chemotherapy. We postulated that cell-cycle kinetics of pre-LSCs may be an important determinant of clonal evolution and therapeutic resistance. Using a doxycycline-inducible H2B-GFP transgene in a mouse model of T-cell acute lymphoblastic leukemia to study cell cycle in vivo, we show that self-renewal, clonal evolution and therapeutic resistance are limited to a rare population of pre-LSCs with restricted cell cycle. We show that proliferative pre-LSCs are unable to return to a cell cycle-restricted state. Cell cycle-restricted pre-LSCs have activation of p53 and its downstream cell-cycle inhibitor p21. Furthermore, absence of p21 leads to proliferation of pre-LSCs, with clonal extinction through loss of asymmetric cell division and terminal differentiation. Thus, inducing proliferation of pre-LSCs represents a promising strategy to increase cure rates for acute leukemia.


Subject(s)
Cell Cycle/genetics , Clonal Evolution/genetics , Leukemia, Myeloid, Acute/genetics , Animals , Cell Cycle/physiology , Clonal Evolution/physiology , Drug Resistance, Neoplasm , Female , Male , Mice , Neoplastic Stem Cells/metabolism , Exome Sequencing/methods
11.
Int J Cancer ; 143(11): 2862-2870, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30183077

ABSTRACT

To investigate tumor clonal evolution in hepatocellular carcinoma (HCC), we collected 31 tumor samples,16 peritumor samples and matched PBMCs from 11 long-term follow-up patients with HCC. Whole-exome sequencing was performed to obtain SNVs and CNVs for each sample. An average of 652.2 somatic mutations were identified in each patient and the mean percentage of nonubiquitous tumor mutations was 63.7% (range, 0.7%-100%), reflecting the variety of tumor heterogeneity. Further analysis of clonal evolution was conducted based on mutation clustering results and revealed that different clonal evolution patterns indeed existed in single and multifocal HCC while these patterns were significantly correlated to patients' clinical course. These patterns clearly demonstrated different mechanisms of tumor recurrence. During tumor clonal evolution, potential therapeutic targets also emerged and vanished dynamically. Moreover, mutation analysis revealed that the contribution of mutational signature was correlated with clonal evolution history. Target sequencing of follow-up plasma samples also confirmed that ctDNA level could dynamically reflect tumor clonal/subclonal burden. By investigating clonal evolution in HCC patients, our analysis revealed that different patterns indeed existed during HCC progression and proposed a novel strategy for identifying the origin of recurrent tumor as well as optimizing treatment selection.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Clonal Evolution/genetics , Clonal Evolution/physiology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , DNA Mutational Analysis/methods , Disease Progression , Exome/genetics , Follow-Up Studies , Humans , Mutation/genetics , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Exome Sequencing
13.
Cancer Res ; 78(10): 2449-2456, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29703720

ABSTRACT

Multiple myeloma (MM) is a treatable, but incurable, malignancy of plasma cells (PC) in the bone marrow (BM). It represents the final stage in a continuum of PC dyscrasias and is consistently preceded by a premalignant phase termed monoclonal gammopathy of undetermined significance (MGUS). The existence of this well-defined premalignant phase provides the opportunity to study clonal evolution of a premalignant condition into overt cancer. Unraveling the mechanisms of malignant transformation of PC could enable early identification of MGUS patients at high risk of progression and may point to novel therapeutic targets, thereby possibly delaying or preventing malignant transformation. The MGUS-to-MM progression requires multiple genomic events and the establishment of a permissive BM microenvironment, although it is generally not clear if the various microenvironmental events are causes or consequences of disease progression. Advances in gene-sequencing techniques and the use of serial paired analyses have allowed for a more specific identification of driver lesions. The challenge in cancer biology is to identify and target those lesions that confer selective advantage and thereby drive evolution of a premalignant clone. Here, we review recent advances in the understanding of malignant transformation of MGUS to MM. Cancer Res; 78(10); 2449-56. ©2018 AACR.


Subject(s)
Cell Transformation, Neoplastic/genetics , Monoclonal Gammopathy of Undetermined Significance/diagnosis , Monoclonal Gammopathy of Undetermined Significance/pathology , Multiple Myeloma/pathology , Plasma Cells/pathology , Precancerous Conditions/pathology , Bone Marrow/pathology , Bone Marrow Cells/pathology , Cell Transformation, Neoplastic/pathology , Clonal Evolution/physiology , DNA Copy Number Variations/genetics , Disease Progression , Humans , Monoclonal Gammopathy of Undetermined Significance/genetics , Multiple Myeloma/diagnosis , Multiple Myeloma/genetics , Mutation/genetics , Tumor Microenvironment/physiology
14.
Int J Hematol ; 108(2): 213-217, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29417354

ABSTRACT

In acute myeloid leukemia (AML), patients may harbor pre-leukemic hematopoietic stem cells (HSCs) containing some, but not all, of the mutations observed in the leukemic cells. These pre-leukemic HSCs may survive induction chemotherapy and contribute to AML relapse by obtaining additional mutations. We report here an acute monoblastic leukemia (AMoL) patient who later developed an unclassifiable myeloproliferative neoplasm (MPN-U). Whole-exome sequencing and cluster analysis demonstrated the presence of three distinct major clones during the clinical course: (1) an AMoL clone with ASXL1, CBL, and NPM1 somatic mutations, likely associated with the pathogenesis, and GATA2, SRSF2, and TET2 mutations, (2) an AMoL remission clone, with mutated GATA2, SRSF2, and TET2 only (possibly the founding clone (pre-leukemic HSC) that survived chemotherapy), (3) a small subclone which had JAK2 mutation during the AMoL remission, appearing at MPN-U manifestation with additional mutations. These findings suggest that pre-leukemic HSCs in AML patients may give rise to non-AML myeloid malignancies. This is the first report to analyze the clonal evolution from AMoL to MPN-U, which may provide new insight into the development of myeloid malignancies.


Subject(s)
Clonal Evolution/genetics , Leukemia, Monocytic, Acute/genetics , Leukemia, Monocytic, Acute/pathology , Myeloproliferative Disorders/etiology , Myeloproliferative Disorders/genetics , Aged , Clonal Evolution/physiology , Hematopoietic Stem Cells/pathology , Humans , Induction Chemotherapy , Leukemia, Monocytic, Acute/blood , Leukemia, Monocytic, Acute/complications , Male , Mutation , Neoplasm Recurrence, Local , Nucleophosmin , Exome Sequencing
15.
PLoS Pathog ; 14(2): e1006861, 2018 02.
Article in English | MEDLINE | ID: mdl-29444188

ABSTRACT

Adult T-cell leukaemia/lymphoma (ATL) arises from chronic non-malignant human T lymphotropic virus type-1 (HTLV-1) infection which is characterized by high plasma pro-inflammatory cytokines whereas ATL is characterized by high plasma anti-inflammatory (IL-10) concentrations. The poor prognosis of ATL is partly ascribed to disease-associated immune suppression. ATL cells have a CD4+CCR4+CD26-CD7- immunophenotype but infected cells with this immunophenotype ('ATL-like' cells) are also present in non-malignant HTLV-1 infection. We hypothesized that 'ATL-like' and ATL cells have distinct cytokine producing capacity and a switch in the cytokines produced occurs during leukemogenesis. Seventeen asymptomatic carriers (ACs), 28 patients with HTLV-1-associated myelopathy (HAM) and 28 with ATL were studied. Plasma IL-10 concentration and the absolute frequency of IL-10-producing CD4+ T cells were significantly higher in patients with ATL compared to AC. IL-10-producing ATL cells were significantly more frequent than 'ATL-like' cells. The cytokine-producing cells were only a small fraction of ATL cells. Clonality analysis revealed that even in patients with ATL the ATL cells were composed not only of a single dominant clone (putative ATL cells) but also tens of non-dominant infected clones ('ATL-like' cells). The frequency of cytokine-producing cells showed a strong inverse correlation with the relative abundance of the largest clone in ATL cells suggesting that the putative ATL cells were cytokine non-producing and that the 'ATL-like' cells were the primary cytokine producers. These findings were confirmed by RNAseq with cytokine mRNA expression in ATL cells in patients with ATL (confirmed to be composed of both putative ATL and 'ATL-like' cells by TCR analysis) significantly lower compared to 'ATL-like' cells in patients with non-malignant HTLV-1 infection (confirmed to be composed of hundreds of non-dominant clones by TCR analysis). A significant inverse correlation between the relative abundance of the largest clone and cytokine mRNA expression was also confirmed. Finally, 'ATL-like' cells produced less pro- and more anti-inflammatory cytokines than non 'ATL-like' CD4+ cells (which are predominantly HTLV uninfected). In summary, HTLV-1 infection of CD4+ T cells is associated with a change in cytokine producing capacity and dominant malignant clonal growth is associated with loss of cytokine producing capacity. Non-dominant clones with 'ATL-like' cells contribute to plasma cytokine profile in patients with non-malignant HTLV-1 infection and are also present in patient with ATL.


Subject(s)
Cell Transformation, Viral/physiology , Cytokines/metabolism , HTLV-I Infections/immunology , HTLV-I Infections/metabolism , Human T-lymphotropic virus 1/physiology , Leukemia-Lymphoma, Adult T-Cell/virology , Aged , Aged, 80 and over , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Clonal Evolution/physiology , Cohort Studies , Cytokines/blood , Cytokines/genetics , Disease Progression , Female , HTLV-I Infections/pathology , Human T-lymphotropic virus 1/immunology , Human T-lymphotropic virus 1/pathogenicity , Humans , Immunophenotyping , Leukemia-Lymphoma, Adult T-Cell/immunology , Leukemia-Lymphoma, Adult T-Cell/metabolism , Male , Middle Aged , Paraparesis, Tropical Spastic/immunology , Paraparesis, Tropical Spastic/metabolism , Paraparesis, Tropical Spastic/pathology , Paraparesis, Tropical Spastic/virology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Viral Load
16.
Blood ; 131(5): 496-504, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29141946

ABSTRACT

Age-related alterations in the human blood system occur in B cells, T cells, cells of the innate system, as well as hematopoietic stem and progenitor cells (HSPCs). Interestingly, age-related, reduced genetic diversity can be identified at the stem cell level and also independently in B cells and T cells. This reduced diversity is most probably related to somatic mutations or to changes in the microenvironmental niche. Either process can select for specific clones or cause repeated evolutionary bottlenecks. This review discusses the age-related clonal expansions in the human HSPC pool, which was termed in the past age-related clonal hematopoiesis (ARCH). ARCH is defined as the gradual, clonal expansion of HSPCs carrying specific, disruptive, and recurrent genetic variants, in individuals without clear diagnosis of hematological malignancies. ARCH is associated not just with chronological aging but also with several other, age-related pathological conditions, including inflammation, vascular diseases, cancer mortality, and high risk for hematological malignancies. Although it remains unclear whether ARCH is a marker of aging or plays an active role in these various pathophysiologies, it is suggested here that treating or even preventing ARCH may prove to be beneficial for human health. This review also describes a decision tree for the diagnosis and follow-up for ARCH in a research setting.


Subject(s)
Aging/physiology , Clonal Evolution/physiology , Hematopoiesis/physiology , Aging/blood , Animals , Clone Cells/physiology , Genetic Variation , Hematologic Neoplasms/blood , Hematologic Neoplasms/genetics , Hematopoietic Stem Cells/physiology , Humans
17.
Rev Invest Clin ; 69(2): 66-76, 2017.
Article in English | MEDLINE | ID: mdl-28453505

ABSTRACT

In recent years, it has become evident that intra-tumor heterogeneity of breast cancer is a big challenge for the diagnosis, treatment, and clinical course of tumor-bearing patients. The advances in molecular biology and other technologies have led to the knowledge that a breast cancer tumor is comprised of multiple cellular entities. Here we review the two theories that have been described, trying to explain the origin of intra-tumor heterogeneity: clonal evolution and cancer stem cells. The first one considers that a single cell gives rise to many subpopulations through the accumulation of multiple aberrations, while the cancer stem cells theory foresees a hierarchical tumor evolution where only a few cells with self-renewal capacity give rise to different subpopulations. We also analyze the genetic, epigenetic, and microenvironment contributions to breast cancer intra-tumor heterogeneity. Finally, the clinical and therapeutic impact of intra-tumor heterogeneity on the outcome of breast cancer patients is discussed.


Subject(s)
Breast Neoplasms/pathology , Clonal Evolution/physiology , Neoplastic Stem Cells/cytology , Breast Neoplasms/diagnosis , Breast Neoplasms/therapy , Cell Self Renewal/physiology , Epigenesis, Genetic/physiology , Female , Humans , Molecular Biology/methods , Tumor Microenvironment/physiology
18.
Rinsho Ketsueki ; 57(2): 118-28, 2016 Feb.
Article in Japanese | MEDLINE | ID: mdl-26935629

ABSTRACT

Acute myeloid leukemia (AML) is a form of blood cancer that is characterized by the rapid growth of abnormal myeloid cells. Although the general therapeutic strategy in patients with AML has not changed substantially in more than 30 years, remarkable progress has been achieved in understanding the pathogenesis of AML. Genome-wide analyses have revealed genetic mutations and epigenetic dysregulations that are present in AML cells. Studies of leukemia stem cells have clarified their complex properties and functions in the development of AML, and have also led to the recent identification of pre-leukemic hematopoietic stem cells that undergo clonal evolution in healthy people. Translation of these new findings into the clinical setting is just beginning. This article focuses on recent advances in basic research on the molecular pathogenesis of AML. New strategies under investigation, including epigenetic therapies and immunotherapies, to provide better therapeutic options for AML patients, are also summarized.


Subject(s)
Clonal Evolution/genetics , Gene Expression Regulation, Leukemic/genetics , Genome-Wide Association Study , Leukemia, Myeloid, Acute/genetics , Acute Disease , Animals , Clonal Evolution/physiology , Humans , Leukemia, Myeloid, Acute/diagnosis , Mutation/genetics
19.
Cancer Res ; 76(6): 1348-53, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26719541

ABSTRACT

Although tumor blood vessels have been a major therapeutic target for cancer chemotherapy, little is known regarding the stepwise development of the tumor microenvironment. Here, we use a multicolor Cre-dependent marker system to trace clonality within the tumor microenvironment to show that tumor blood vessels follow a pattern of dynamic clonal evolution. In an advanced melanoma tumor microenvironment, the vast majority of tumor vasculature clones are derived from a common precursor. Quantitative lineage analysis reveals founder clones diminish in frequency and are replaced by subclones as tumors evolve. These tumor-specific blood vessels are characterized by a developmental switch to a more invasive and immunologically silent phenotype. Gene expression profiling and pathway analysis reveals selection for traits promoting upregulation of alternative angiogenic programs such as unregulated HGF-MET signaling and enhanced autocrine signaling through VEGF and PDGF. Furthermore, we show a developmental switch in the expression of functionally significant primary lymphocyte adhesion molecules on tumor endothelium, such as the loss in expression of the mucosal addressin MAdCAM-1, whose counter receptor a4ß7 on lymphocytes controls lymphocyte homing. Changes in adhesive properties on tumor endothelial subclones are accompanied by decreases in expression of lymphocyte chemokines CXCL16, CXCL13, CXCL12, CXCL9, CXCL10, and CCL19. These evolutionary patterns in the expressed genetic program within tumor endothelium will have both a quantitative and functional impact on lymphocyte distribution that may well influence tumor immune function and underlie escape mechanisms from current antiangiogenic pharmacotherapies.


Subject(s)
Clonal Evolution/physiology , Endothelium, Vascular/physiology , Lymphocytes/pathology , Tumor Escape/physiology , Animals , Cell Adhesion/physiology , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Chemokine CCL19/metabolism , Chemokines, CXC/metabolism , Endothelium, Vascular/metabolism , Gene Expression Profiling/methods , Hepatocyte Growth Factor/metabolism , Lymphocytes/metabolism , Mice , Mucoproteins , Neoplasms/metabolism , Neoplasms/pathology , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction/physiology , Tumor Microenvironment/physiology , Up-Regulation/physiology
20.
Crit Rev Oncol Hematol ; 98: 116-21, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26589397

ABSTRACT

Multiple myeloma (MM) is composed of an array of multiple clones, each potentially associated with different clinical behavior. Previous studies focused on clinical implication of centrosome amplification (CA) in MM show contradictory results. It seems that the role of CA as well as CA formation in MM differ from other malignancies. This has brought about a question about the role of CA positive clone which is--is it going to be a more aggressive clone evolutionally arising under pressure of negative conditions or can CA serve as a marker of cell abnormality and lead to cell death and further elimination of this damaged subpopulation? This current review is devoted to the discussion of the existence of MM subclones with centrosome amplification (CA), its evolutionary behaviour within intraclonal heterogeneity as well as its potential impact on the disease progression and MM treatment.


Subject(s)
Centrosome/pathology , Clonal Evolution/physiology , Multiple Myeloma/pathology , Apoptosis/physiology , Carcinogenesis/pathology , Cell Proliferation , Clone Cells , Disease Progression , Humans , Multiple Myeloma/genetics , Stress, Physiological/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...